19

Full Issue ID Full Issue... · 2021. 1. 28. · She had to wear a full gown and mask, and never once complained. We have been together for seven years, and over half of that has been

  • Upload
    others

  • View
    0

  • Download
    0

Embed Size (px)

Citation preview

  • yale cancer center

    Peter G. Schulam, MD, PhD

    Interim Director

    Kevin Vest, MBA, FACHE

    Deputy Director, Administration and Clinical Affairs

    Renee Gaudette

    Director, Public Affairs and Marketing

    art + design

    Peter Baker Studios LLC

    pbakerstudios.com

    contributorsWriters

    Emily Fenton

    Steve Kemper

    Photographer

    Peter Baker

    pbakerphoto.com

    Breakthroughs is published annually to highlight research and clinical advances from

    Yale Cancer Center and Smilow Cancer Hospital at Yale-New Haven.

    Yale Cancer Center

    333 Cedar Street, PO Box 208028

    New Haven, CT 06520-8028

    yalecancercenter.org

    © Copyright 2015, Yale Cancer Center.

    All rights reserved. No part of this periodical may be reproduced by any means,

    prints, electronic, or any other, without prior written permission of the publisher.

    Editorial Office

    Yale Cancer Center

    100 Church Street South, Suite 160

    New Haven, CT 06519

    [email protected]

    On the CoverDr. Andrea Silber with Maria Castellani,

    Wendy Ormond, and Marta Vega

    2 Director’s Letter

    features 4 Detecting Hereditary Kidney Cancer

    Once assumed a sporadic disease, newer

    research suggests that five to eight percent of

    kidney cancer is genetic and Dr. Brian Shuch,

    director of Smilow Cancer Hospital’s Hereditary

    Kidney Cancer Program believes this number

    will continue to rise.

    7 Bringing Clinical Trials into our Communities

    Dr. Andrea Silber’s close relationship with her patients and growing network in the New Haven community is building trust and expanding clinical trial participation through a new initiative called OWN IT.

    10 Accelerating Development of New Drugs

    The new Yale Cancer Therapeutic Accelerator Program (Yale CTAP) will focus on generating new cancer drugs and accelerating their development in much the same way as biotech startups.

    12 Love Knows no Bounds, Not Even Cancer

    Josh Scussell was diagnosed with T Cell Lymphoma at 23. His long road to cure included two transplants and several years. Throughout, Heather was by his side and they married one year and three months after his final transplant.

    research programs 16 Radiobiology and Radiotherapy

    Imaging the Epidermal Growth Factor Receptor

    18 Cancer Prevention and ControlTexting to Check in on Patients

    20 Cancer ImmunologyThe Immune System and the Risk of Lymphoma

    22 Virus and Other Infection-associated Cancers

    The Links Between Typhoid, Bacteria, and Cancer

    24 Developmental TherapeuticsMolecules That Seek and Destroy Mutant Proteins

    26 Cancer Genomics, Genetics, and EpigeneticsBioinformatics Boosts Cancer Research

    28 Signal TransductionDecoding the Signals of Metastasis

    leadership and membership 30 Yale Cancer Center and

    Smilow Cancer Hospital at Yale-New Haven Leadership

    32 Yale Cancer Center Membership

    1yalecancercenter.org | Yale Cancer Center

    10

    12

    7

  • Over the last year, Yale Cancer Center and Smilow

    Cancer Hospital at Yale-New Haven have focused

    greater emphasis on the need to provide education and

    community-based outreach initiatives that are important

    to our patients. Yale Cancer Center and Smilow Cancer

    Hospital recognize the importance of impacting the

    community we serve – the communities in which our

    patients as well as our physicians, faculty, scientists, and

    employees share. New Haven and its surrounding towns

    provide us with an opportunity to care for and educate

    a diverse population on cancer prevention, screening,

    and treatment.

    In the months ahead, we will implement a highly

    organized effort to expand cancer screening in diverse

    populations, particularly in men and women at the

    highest risk of developing certain cancers. We will

    establish important initiatives aimed at increasing

    the opportunity for all patients in our community to

    access the most recent advances in treatment through

    our clinical trials. With support from many of their

    colleagues, Beth Jones, PhD, MPH and Andrea Silber,

    MD are partnering to lead these efforts.

    Our refocused efforts in clinical trial outreach and

    patient navigation continues Dr. Silber’s long tradition of

    patient education in the community of New Haven. In

    2015, Dr. Silber launched OWN IT (Oncologists Welcome

    New Haven Into Trials) to support more minorities as

    they consider participation in clinical trials. New Haven’s

    population is approximately two-thirds African American

    and Latino and we recognize the need for additional

    support for this diverse population during cancer

    treatment. What makes these efforts so crucial, is the

    need to ensure the research that cancer centers do across

    the country reflects the diverse population of the United

    States. Although the national cancer research community

    often falls short of this aspiration, Yale Cancer Center

    is committed to doing our part to help achieve this

    important goal.

    Yale Cancer Center also continues to expand new

    scientific programs and recently launched the Yale

    Cancer Therapeutic Accelerator Program (Yale CTAP)

    to improve our ability to discover compounds that may

    be able to be rapidly converted to new treatments for

    patients. This new program, led by Craig Crews, PhD

    and Mark Lemmon, PhD, will facilitate the advancement

    of these compounds or drugs from Yale Cancer Center

    laboratories. The program helps to provide Yale Cancer

    Center drug discovery teams with the support needed

    to increase the likelihood of commercialization of those

    compounds. I look forward to sharing some of the

    success stories from Yale CTAP over the coming year as

    Dr. Crews and Dr. Lemmon begin to build the team and

    infrastructure needed to optimize drug discovery at Yale

    Cancer Center.

    The momentum developed over the first 5 years at

    Smilow Cancer Hospital continued to expand in 2015.

    In June, we welcomed Saint Francis Hospital in Hartford

    to our Smilow Cancer Care Network and have expanded

    access to Yale Cancer Center physicians along the eastern

    shoreline from Guilford to Old Saybrook, and most

    recently to Waterford. Exceptional care and access to

    clinical trials is now available at 12 locations around the

    state of Connecticut. Yale’s clinical trial accrual also

    reached an all-time high in 2015 and we continue to strive

    to make the newest treatment options available to our

    patients through clinical trial participation.

    In addition, Yale Cancer Center’s 7 Research Programs

    and nearly 350 members are continuing their pursuit to

    understand cancer and find ways to reduce its impact on

    patients in our community. New discoveries happen daily

    as scientists from our laboratories and physicians from

    our Disease Aligned Research Teams (DARTs) at Smilow

    Cancer Hospital combine their efforts in unique and

    impactful ways. Many highlights from 2015 are featured

    in this edition of Breakthroughs and I look forward to

    sharing new research advances and outcomes from our

    laboratories and clinics with you in the year to come.

    Sincerely,

    Peter G. Schulam, MD, PhD

    Director, Yale Cancer Center and Physician-in-Chief,

    Smilow Cancer Hospital at Yale-New Haven (Interim)

    3yalecancercenter.org | Yale Cancer Center

  • Brian Shuch, MD, Assistant

    Professor of Urology and of

    Radio logy and Biomedica l

    Imaging, has a patient in her 70s who

    probably wouldn’t be alive if not for the

    Hereditary Kidney Cancer Program, which

    Dr. Shuch directs. Before Dr. Shuch saw her,

    she had lost a kidney to cancer and wasn’t

    responding to standard chemotherapy. Her

    doctor referred her to Dr. Shuch’s program,

    which specializes in kidney cancers caused by

    heredity or acquired genetic mutations.

    As always, Dr. Shuch delved into her

    family history. Her mother had died of kidney

    cancer. So had both of her own children, who

    hadn’t responded to standard treatment of

    the disease. Dr. Shuch suspected an inherited

    syndrome. A genetic test confirmed it: she had

    a variant of an uncommon kidney cancer called

    4

    Brian Shuch, MD

    5yalecancercenter.org | Yale Cancer Center

    hereditary leiomyomatosis and renal cell cancer (HLRCC). He put her on

    an experimental regimen of chemotherapy not typically used against kidney

    cancer, but known to be effective against HLRCC. A year later, her cancer had

    essentially disappeared.

    Smilow Cancer Hospital launched the Hereditary Kidney Cancer Program in 2013 precisely to

    help such patients and to educate the physicians who diagnose them. Kidney cancer is one of the most common

    forms of the disease, with an estimated 61,500 new cases each year. Scientists once assumed that kidney cancers occur

    sporadically, but newer research suggests that five to eight percent stem from genetic predisposition. Dr. Shuch believes

    this number will continue to rise.

    “We haven’t even begun to scratch the surface of inherited predisposition,” he said. “Lineage studies show that about

    80 percent of kidney cancers cluster in about a quarter of the population, strong evidence that kidney cancer has a strong

    hereditary basis.”

    Currently, however, genetic syndromes associated with kidney cancer often go unrecognized by physicians, to the

    detriment of patients. The standard of care used against sporadic forms of the disease may be ineffective against hereditary

    syndromes. Certain syndromes are associated with asymptomatic signs of kidney cancer. Four syndromes, for example,

    can manifest themselves as skin problems. Few physicians have the training to recognize these signs. Similarly, a syndrome

    called Von Hippel-Lindau syndrome may be accompanied by retinal tumors.

    “I’ve had patients lose their vision by a delay in the diagnosis of manifestations outside the kidney,” said Dr. Shuch.

    “A urologist and oncologist who are focused on their areas of expertise often miss subtle signs in the physical exam or

    cues in the

    family history

    t h a t w i l l r a i s e

    alarms to an experienced

    multidisciplinary team.” The

    team at Yale includes urologists, pathologists,

    dermatologists, geneticists, and genetic counselors.

    Just a day earlier, Dr. Shuch had seen a patient referred

    to him by the team’s dermatologist because of a skin

    lesion called a leiomyoma, sometimes a sign of HLRCC.

    When Dr. Shuch teased out the patient’s family history, he

    learned that all three of his sisters had had their uteruses

    removed in their 30s because of fibroids, another sign

    of HLRCC, and his 23-year-old nephew had advanced

    kidney cancer.

    “That patient doesn’t have any symptoms of kidney

    cancer,” said Dr. Shuch, “but he absolutely needs testing

    and genetic screening for a presumed diagnosis of

    HLRCC. We’ve had patients whose lives were potentially

    detecting hereditary KIDNEY CANCER

  • saved by identifying an asymptomatic kidney cancer

    through screening and treating it before dissemination.”

    Part of Dr. Shuch’s mission is to educate physicians to

    take a comprehensive family history, and to be alert for

    signs that a patient should be referred to his program for

    evaluation and genetic testing. One of the clearest signs

    is early onset of kidney cancer. The average age for a

    diagnosis is 64, and the cancer rarely hits people younger

    than 45 – but the inherited forms often strike before that

    age. Other signs: unusual pathology, bilateral disease, and

    multifocal disease.

    Dr. Shuch and his colleagues have developed a genetic

    screening panel for the known syndromes, which at this

    point number about 15. Yale is one of the first centers

    with such a panel, which allows the team to test multiple

    genes at once.

    Genetic testing is crucial not only for patients with

    symptoms of hereditary kidney cancer, but for their

    parents, siblings, and children. Early detection can

    mean the difference between life and death. HLRCC,

    for instance, is one of the most aggressive syndromes.

    Before Dr. Shuch’s program began, Yale had not

    identified a single patient with HLRCC, but in the past

    two years, nine families have been diagnosed with it.

    “Rather than lumping them in with generic kidney

    cancer and treating them the same,” explained Dr.

    Shuch, “we have been able to offer them unique treatment strategies focused on the genetic alterations associated with

    that syndrome.”

    That has prolonged lives. James Uhl, for instance, was 45 when diagnosed with kidney cancer. The disease had spread

    to his liver, lungs, bones, brain, eyes, and lymph system. He was referred to Dr. Shuch, who found HLRCC. In January

    2014, Dr. Shuch removed Mr. Uhl’s left kidney, part of his liver, and multiple lymph nodes, then put him on a unique

    regimen not designed for kidney cancer but often effective against this particular syndrome. It worked well for six

    months, an unusual span against a cancer so advanced. After trying several other regimens, in late 2015 Dr. Shuch got

    Mr. Uhl into a clinical trial at the NCI. Dr. Shuch doubts that Mr. Uhl would have had the last two years with his family

    without the Smilow program.

    “Brian’s knowledge of the disease has offered me tremendous guidance,” said Mr. Uhl, who remains optimistic.

    “Without the work being done by him and his team, little would be known about this aggressive disease. Because of

    their research, more and more treatment options are going into trial phases, which gives patients like me a lot of hope.”

    He’s also grateful that Dr. Shuch identified his syndrome, because his children, 10 and 8, can be tested very early for the

    gene. “I’ll be able to potentially save my children’s lives,” he said.

    As knowledge about the genetics associated with each kidney cancer syndrome grows, targeted therapies become

    possible. “Rather than treating kidney cancer as one disease,” said Dr. Shuch, “we’ve learned it’s a group of cancers

    arising from the same organ, but all very different.”

    New knowledge is changing treatment in other ways as well. For instance, three of the syndromes produce kidney

    cancers that pose no risk of metastasis until the tumor reaches a certain threshold. Until then, the cancer can be

    managed, and patients can be spared unnecessary surgery—that is, if the syndromes are identified early, which is why

    testing can be so important.

    In 2016 the program will expand to other urologic cancers with heritable syndromes such as prostate, bladder, and

    testicular cancers. Dr. Shuch and his colleague are also planning a tele-genetic counseling service so that other cancer

    centers can benefit from Yale’s expertise.

    “So much of this is not just cancer treatment,” explained Dr. Shuch, “it’s preventing the delayed management of new

    tumors by aggressive screening. A delay in diagnosis could be the difference between cure and cancer dissemination.”

    6 Yale Cancer Center | Year in Review 20158 Yale Cancer Center | Year in Review 2014

    Henry Baker

    PAVING THE WAY FOR THE FUTURE:Tale of TriumphHENRY BAKER’S Communitiesinto our

    Andrea Silber, MD

    Andrea Silber, MD, Associate Clinical

    Professor of Medicine, has never forgotten one of

    her first patients in New Haven nearly thirty years ago, a

    young black mother of four. She had breast cancer but had

    never been treated. Within a week, she was dead.

    “It was incredibly sad, and it stuck with me,” said Dr.

    Silber, a medical oncologist who specializes in breast

    cancer. “I was also struck by the fact that in New Haven,

    whenever you see people of color with cancer, it’s worse

    in so many ways. They may come in later, or the onset

    may come at a much younger age, or they may have other

    chronic conditions that make it difficult to treat their

    cancer. I’ve had a long interest in decreasing cancer health

    7yalecancercenter.org | Yale Cancer Center

    Bringing CLINICAL TRIALS

  • 8 Yale Cancer Center | Year in Review 2015

    disparities in New Haven.”

    For decades these disparities didn’t get much attention

    from the medical establishment. That has recently started

    to change, spurred partly by the Affordable Care Act. Dr.

    Silber is determined to add to the momentum.

    One of her tools is a new program called OWN IT

    (Oncologists Welcome NewHaven Into Trials). OWN

    IT’s purpose is to get more minorities from inner city

    New Haven into clinical trials for new cancer treatments

    conducted at Smilow Cancer Hospital. Minorities are

    under-represented in such trials nationwide. The city of

    New Haven, for instance, is about two-thirds black and

    Latino, yet those groups account for about 12 percent of the

    participants in Yale’s cancer clinical trials. (The trials that

    Dr. Silber accrues to are more than 50 percent minority.)

    One of OWN IT’s tasks is to remove the barriers

    keeping minorities out of trials. The first barrier is

    mistrust. “People in town have been afraid of being

    someone’s research project,” said Dr. Silber. “They say,

    ‘Why should I do this? Who is this going to help? I don’t

    want to be experimented upon.’” To overcome these

    suspicions and build trust, says Dr. Silber, physicians

    must become more patient and communicate more

    clearly. “Everyone has a right to understand what’s

    happening to their bodies, and it’s my obligation to

    explain it in a way they understand.”

    That’s how Dr. Silber convinces her patients to enter

    clinical trials at Smilow. Maria Castellani, a mother of

    three, had a bout with stage 3 breast cancer in 2011 and

    survived, but in June 2014 the disease returned, in the

    aggressive triple negative form. It spread to her lungs,

    lymph nodes, and bones. She saw four oncologists, one

    of whom gave her six months to live. Three of them never

    mentioned clinical trials. The fourth suggested a trial, but

    instead of explaining it, merely handed Ms. Castellani a

    thick packet of baffling technical information.

    Distraught and despairing, she finally connected with

    According to Dr. Silber, these low numbers have

    consequences. First, minorities are missing out on the most

    cutting-edge treatments for their cancers, which affects their

    survival rates. Second, the lack of minorities can skew trial

    results – Dr. Silber has noticed that new medications given

    to her patients often don’t live up to their hype.

    “So I would go back and look at the trial,” she said,

    “and I realized that my patient never would have been

    chosen for the study, because it eliminated anyone with

    complicating factors like diabetes or high blood pressure

    or obesity, which are common among my cancer patients

    from the inner city. If you test all your oncologic drugs

    only on the healthy, the wealthy, and the wise, your

    test doesn’t equate to real life and doesn’t address the

    problems we see in New Haven.”

    “I’m so grateful for being in this clinical trial,” Ms. Castellani said. “Dr. Silber

    was the only doctor who didn’t give up on me. She took her time and gave me

    this option, and she explained everything.”

    9yalecancercenter.org | Yale Cancer Center

    the clinical trial.” During treatment, Dr. Silber asked her

    to speak to two Latina patients with breast cancer who

    were afraid of trials. “I was kind of an advocate,” said Ms.

    Vega, “and I felt good because I was helping someone else

    and telling them it’s OK.”

    After Wendy Ormond was diagnosed with breast cancer,

    she went into a tailspin. She began drinking and contracted

    pancreatitis before she could have surgery, and her blood

    pressure also shot up. She was referred to Dr. Silber, who

    sent her to a heart doctor to get her blood pressure under

    control. She also convinced Ms. Ormond to overcome her

    terror of doctors and enter a clinical trial. “I trusted her,”

    said Ms. Ormond simply. “I knew she wouldn’t get me into

    something that would hurt me. I love that lady.” She began

    the trial in February 2015, and her blood tests continue to

    improve.

    OWN IT will address the barriers of fear and mistrust,

    says Dr. Silber, by building a relationship between Yale

    and the people in the city surrounding it. That will take

    time. She has always devoted time to community outreach,

    but now new funding from the National Institutes of

    Health and other sources will allow her to hire patient

    navigators and a community health educator who can hold

    educational sessions at community centers, schools, and

    churches, to explain clinical trials and their advantages.

    She adds that the city’s primary care physicians and

    Yale oncologists also need educating. Too many patients

    never hear about trials. Patients who don’t speak English

    may be rejected as too time-consuming. Some trials

    disqualify swaths of inner city minorities because their

    blood pressure, cholesterol, or glucose levels tend to be

    higher than those of people in wealthier suburbs.

    However, Dr. Silber sees signs of change. The NIH,

    the National Cancer Institute, and the American Society

    of Clinical Oncology all have started paying attention

    to health disparities and funding research. Things are

    changing at Yale as well. In May, Dr. Silber was appointed

    to the Cancer Center’s Executive Committee as Assistant

    Director for Diversity and Health Equity.

    “We’ve never been at the table before,” said Dr. Silber,

    “but because of the leadership at Smilow, there’s a new

    awareness of how this needs to be part of the thinking.

    Everything is aligning to make OWN IT a way to really

    change clinical trials for inner city New Haven.”

    Dr. Silber, who suggested a clinical trial with Orteronel.

    Ms. Castellani’s mother, brothers, and uncles warned

    that she would be treated like a lab rat, but she trusted Dr.

    Silber and joined the trial in July 2015. By the end of the

    year, her lymph nodes were clear, her lungs were stable,

    and her bone cancer was fading.

    “I’m so grateful for being in this clinical trial,” she said,

    crying a little. “Dr. Silber was the only doctor who didn’t

    give up on me. She took her time and gave me this option,

    and she explained everything. I don’t have enough to pay

    her for what she’s done for me.”

    Similarly, Marta Vega was diagnosed with stage 3

    breast cancer. Her oncologist was hurried and distant.

    Terrified and in denial, she didn’t want to consider

    treatment of any kind. Then she saw Dr. Silber, who

    carefully explained a breast cancer trial to her and said,

    “If you were my sister, I would recommend this.” Her

    warmth and patience persuaded Ms. Vega, who received

    experimental chemotherapy from March to August 2014.

    Her tumor shrank 75 percent, and surgery took care of

    the rest. “If it wasn’t for Dr. Silber,” said Ms. Vega, “I

    wouldn’t have survived, because I would not have done

  • 10 Yale Cancer Center | Year in Review 2015

    Craig Crews, PhD; Peter G. Schulam, MD, PhD; Mark Lemmon, PhD

    The Valley of Death: that’s what the co-directors

    of the new Yale Cancer Therapeutic Accelerator Program

    (Yale CTAP) call the place where promising lab compounds

    perish on the rigorous journey towards becoming a new

    cancer drug. In fact, most patents of discoveries expire

    before they even reach the valley’s outskirts, according

    to Craig Crews, PhD, Lewis B. Cullman Professor of

    Molecular, Cellular and Developmental Biology, and

    Director of the Yale Center for Molecular Discovery, and his

    CTAP co-director Mark Lemmon, PhD, David A. Sackler

    Professor of Pharmacology and co-director of the Cancer

    Biology Institute.

    “Despite all the efforts at translational medicine,” said

    Dr. Crews, “the harsh reality is that it takes a lot to develop

    a tool compound into a drug candidate. There’s still a gap,

    the proverbial Valley of Death. An academic can take his

    or her research only so far towards translation, because

    there aren’t funding mechanisms for it, there isn’t the

    training for it, there isn’t the personnel to do it.”

    “Basic scientists in the Cancer Center are doing

    amazing work,” added Dr. Lemmon, “but there are

    11yalecancercenter.org | Yale Cancer Center

    NEW DRUGSof NEW DRUGSothers to ask what it would take to make something

    attractive enough to put $5 million or $15 million into a

    new startup located here,” said Dr. Crews. “So we really

    aren’t doing drug discovery or drug development, but

    would generate the companies to do that.”

    Another part of the program’s mission is to teach YCC

    members how to be more entrepreneurial. Once the

    external advisory committee approves a project, CTAP will

    work with the initiating faculty member to put together

    a business plan and coach him/her on how to pitch the

    discovery to venture capitalists.

    Because the program is so new, the search for funding

    has just begun. Dr. Crews and Dr. Lemmon intend to

    tap philanthropic and commercial sources that prefer to

    invest in projects that have moved beyond basic science

    toward practical application. A strong selling point, said

    the co-directors, is that CTAP can draw on new state-of-

    the-art research facilities and Institutes on Yale’s West

    Campus as well as several departments.

    “Investors in these new drugs will be getting a lot of

    added value here,” said Dr. Lemmon. “It takes a village

    to move from a germ of an idea to the exploitation of

    the idea.”

    huge barriers to taking their discoveries beyond the lab.

    Expense and lack of access to the right experience stall

    most potential cancer care discoveries. CTAP would

    provide stepping stones across the Valley of Death.”

    The idea for CTAP came from Peter G. Schulam, MD,

    PhD, Director of Yale Cancer Center and Physician-

    in-Chief of Smilow Cancer Hospital at Yale-New

    Haven (Interim). The program is modeled on a similar

    one started by Dr. Crews called PITCH (Program in

    Innovative Therapeutics for Connecticut’s Health), which

    is also aimed at developing new drugs. PITCH received a

    $10 million, three-year grant from the state of Connecticut

    to select and fund projects from Yale and the University

    of Connecticut, with the goal of developing promising

    discoveries to the point where they can be pitched to

    venture capital firms.

    CTAP will adopt this concept, but with a focus on

    generating new cancer drugs and accelerating their

    development in much the same way as biotech startups.

    “By coalescing ideas, capital, and talent,” said Dr. Crews,

    “one can drive the whole drug discovery process in a much

    faster way than the pharmaceutical industry has.” CTAP

    will bring together programs across multiple schools

    and faculties at Yale to leverage their unique strengths in

    this process, allowing Cancer Center members to work

    alongside faculty who can complement their research goals

    from throughout the University.

    It would work like this: let’s say a Cancer Center

    member makes an intriguing discovery and wants to

    develop it into a drug that could be tested in Phase I trials at

    Yale, but doesn’t know how to go about it. CTAP and the

    Yale Center for Molecular Discovery would engage a team

    of professional drug developers to screen drug candidates

    for useful compounds. If any of these appeared promising,

    medicinal chemists and others would design, synthesize,

    and tweak the compounds to make them more drug-like.

    CTAP would then help marshal the efforts of labs in the

    Cancer Biology Institute and elsewhere to generate data

    packages evaluating the potential of the compounds –

    facilitating the pipeline from idea to therapeutic approach.

    At that point, the compounds would be evaluated by an

    external advisory board, comprised of representatives

    from biotech and big pharma, to determine which have

    therapeutic potential and should advance to the next level:

    pitching to philanthropists, venture capitalists, and others.

    “We want to work with the venture capitalists and

    developmentACCELERATINGACCELERATING

  • At the age of 23, Josh Scussell was starting his life,

    and like most people in their early twenties, cancer was

    the last thing on his mind. He was still on his parent’s

    insurance and did not have a primary care doctor.

    Therefore, when he noticed a lump on his left leg in

    the groin area, he went to a walk-in clinic to have it

    examined. Thinking it was an abscess, the lesion was

    lanced, but when blood, not puss, came out, and Josh’s

    leg immediately became red and inflamed, he was

    instructed to go to a local Emergency Department.

    From that point on, Josh would encounter a lot of

    waiting. He was in the ED from 6PM until 9AM waiting

    for a room before he could be seen, and from there he

    was tested for everything from cat scratch fever to other

    infections and eventually a biopsy was performed. During

    the biopsy, fluid developed around Josh’s heart and he

    went into congestive heart failure. After spending three

    12 Yale Cancer Center | Year in Review 2015

    Josh and Heather Scussell

    13yalecancercenter.org | Yale Cancer Center

    days in the ICU his heart rate and oxygen levels returned to normal. His girlfriend at the

    time, Heather, now his wife, was by his side throughout the entire endeavor.

    “Heather’s mother, who is a nurse, was the first to bring up the possibility of cancer,”

    said Josh. “The doctors assured us this was not the case and that a biopsy would confirm

    this. Once I was out of the ICU and recovering, I received the results of the biopsy, which

    determined I had Non-Hodgkins T Cell Lymphoma. I was in shock and completely

    devastated. I was alone in my room at the time, and when my mother came in and

    heard the news she was more in shock than I was. It was a very emotional time

    for everyone.”

    Josh was started on a chemotherapy regimen know as CHOP, an aggressive form of

    chemotherapy. An earlier PET scan had revealed the disease had spread to Josh’s lymph

    nodes and a large tumor in his groin area. After 6 cycles of treatment with CHOP, Josh’s

    PET scan was completely clear. Josh was relieved and was ready to put it all behind him;

    however, he soon learned that without a stem cell transplant, the cancer could return

    within three months. At this point, it was suggested that he transfer his care and meet

    with one of the top T Cell Lymphoma doctors in the country, Dr. Francine Foss at

    Smilow Cancer Hospital.

    “Dr. Foss got right down to business and explained the next steps in the process, which

    helped me keep the momentum going forward,” explained Josh. “I had already been

    through a lot, and wasn’t sure I could face a stem cell transplant.”

    Just six months since Josh first noticed the lump, he underwent his first transplant,

    an autologous transplant where his own stem cells were collected before treatment and

    then returned to replace the stem cells that had been damaged by the chemotherapy. He

    spent three weeks in the hospital and experienced difficult side effects, but despite this, he

    recovered quickly and was back to his normal life after about three months.

    A year later, Josh began having back pain. When the pain did not lessen, Dr. Foss ordered

    a PET scan and MRI, which revealed a spot that was soon biopsied. The results indicated the

    cancer had returned, this time on his T10 vertebrae. “Had we waited any longer,” said Josh “it

    would have spread to my spine and this would be a different story.”

    For his second transplant, Josh receive an allogeneic transplant along with a new

    chemotherapy regimen that targeted a specific protein in the lymph cells, Brentuximab.

    An allogeneic stem cell transplantation involves transferring stem cells from a healthy

    donor match after chemotherapy in patients where their cancer has relapsed. Dr. Foss

    commented, “Josh’s story is a tremendous success story for the use of newer therapies.

    Love Knows no Bounds,Not Even Cancer

  • When patients relapse after autologous therapies, it’s often difficult to go on with an

    allogeneic transplant because using conventional chemotherapy, the cancer often does

    not go into remission. Because Josh did so well with the new drug, Brentuximab, he was

    the perfect candidate to go ahead with an allogeneic transplant.”

    Six months passed before a donor match was found, but unfortunately the donor

    decided at the last minute not to go through with stem cell donation for Josh. The search

    continued using Be the Match® and Josh received word that a 25-year-old female in

    Canada had been located as a near perfect match for him. His transplant was scheduled

    two years after his first. Due to insurance issues, the transplant was done in Boston at Dana

    Farber. Dr. Foss was kept informed every step of the way. Josh was in the hospital for three

    weeks undergoing chemotherapy before the transplant and during this time, Heather spent

    every night by his side.

    “I couldn’t imagine having to see her go through something like this. She has been with

    me from that first night in the ED up until now,” said Josh. “She was commuting from

    Boston to Guilford, CT every day for almost a month. She had to wear a full gown and

    mask, and never once complained. We have been together for seven years, and over half of

    that has been dealing with this cancer. I knew after that, she would always be by my side,

    and so I asked her if I could spend the rest of my life by hers. She said yes.”

    Following the transplant, due to the high risk of infection, Josh had to undergo one

    year of isolation where he couldn’t leave the house. He lost 50 pounds and recovery was

    slower this time around. A year post transplant, Josh was eager to contact his donor,

    the 25-year-old woman from Canada. As it turns out, they had more in common than

    just bone marrow. Their birthdays are a month apart, and both got married last year.

    Additionally, from Saskatchewan, her husband, a professional hockey player, is a family

    friend of Mandi Schwartz, a former Yale University ice hockey player that passed away

    from acute myeloid leukemia in 2011. For five years, Yale University has been helping save

    lives through marrow donor registration drives in honor of Mandi, including one that was

    held in Mandi’s hometown, Saskatchewan, Canada, where Josh’s donor volunteered and

    decided to join the registry.

    “Her husband’s best friend had leukemia and did not survive his transplant, so when

    they did not hear from me after six months, they thought I had not survived either,” said

    Josh. “I was able to receive her contact information after one year, and they were very

    happy to hear from me. My first email to her was titled ‘You saved my life’ and from there

    we began exchanging photos and information about each other. One of the photos she

    sent was of her on donation day with the bag of stem cells. It was flown in that day and I

    remember seeing the bag and touching it. And there she was in a gown, holding the bag of

    cells and smiling. It was very emotional for me to see that.”

    This past October, one year and three months after his second transplant, Josh and

    Heather were married and honeymooned in Disney World. “I was really excited to hear

    that Josh and Heather got married. He has been a tremendous success story and I am

    “I was really excited to hear that Josh and Heather got married. He has been a tremendous success story and

    I am so happy for them.” - Dr. Francine Foss

    14 Yale Cancer Center | Year in Review 2015 7yalecancercenter.org | Yale Cancer Center

    so happy for them. It was so exciting to see the picture of

    Josh and his bride at their wedding – I felt like it was my

    son getting married,” Dr. Foss said.

    “It truly meant a lot to see how genuinely happy Dr.

    Foss was for me when I got married,” said Josh. “She

    helped me through this difficult time, by more than just

    treating me, but by supporting me.”

    Josh and Heather are looking forward to a long life

    together. They will never forget the woman that saved his

    life and plan to meet her and her husband this spring. Her

    small gesture in joining the bone marrow registry created a

    big connection spanning thousands of miles, and allowed

    for Josh and Heather’s happy ending to be possible.

    “I knew after that, she would

    always be by my side, and so I

    asked her if I could spend the rest

    of my life by hers. She said yes.”

    15yalecancercenter.org | Yale Cancer Center

  • Yale scientists have pioneered a new form of

    positron emission tomography (PET). This new approach

    to imaging may allow cancer-care specialists to detect the

    epidermal growth factor receptor (EGFR), a target for

    cancer therapy that is expressed on the surface of tumor

    cells. This new type of PET scan will also monitor drugs

    that target the EGFR in tumors and could provide a new

    way to image their effectiveness in patients.

    The most common form of PET scan is called

    fluorodeoxyglucose (FDG)-PET, in which a cancer patient

    is given a radioactive sugar that accumulates in the tumor,

    allowing doctors to view it. The Yale group is using PET

    technology in a way that hasn’t been done before. Their

    new technique uses a radiolabeled drug that specifically

    binds to the EGFR. The goal is to provide a method for

    doctors to see the tumor, watch the uptake of the drug,

    and monitor its effectiveness.

    “That’s what we’re really excited about,” said Joseph

    Contessa, MD, PhD, Associate Professor of Therapeutic

    Radiology and Pharmacology whose laboratory studies

    the biology of EGFR. “We have not previously been able

    to non-invasively monitor the interaction of a cancer

    drug with its target in patients. With this work we hope to

    translate our knowledge about the biology of EGFR to a

    simple and clinically useful PET scan.”

    PET imaging of the EGFR is accomplished by using a

    radiotracer called [11C]-erlotinib. Erlotinib is a drug that

    blocks activation of the EGFR, and potently blocks the

    growth of tumor cells with mutation of the EGFR. EGFR

    mutations are found in multiple cancers and come in

    many forms. Those with tyrosine kinase mutations, which

    activate the receptor’s activity, are the Yale team’s current

    focus. The radiolabeled form of erlotinib was planned

    and synthesized by a member of the team, radiochemist

    Yiyun Henry Huang, PhD, Professor of Radiology and

    Biomedical Imaging, and Co-Director of The Yale PET

    Center. Dr. Huang replaced one of the drug’s carbons with

    a radioactive carbon, which allows the compound to be

    traced by PET.

    The team tested the technique by inject ing

    [11C]-erlotinib into mice with lung cancer tumors

    harboring EGFR mutations. Evan Morris, PhD, Associate

    Professor of Radiology and Biomedical Imaging,

    Psychiatry, and Biomedical Engineering, and Co-Director

    of the PET Imaging Section, modeled the radiotracer’s

    distribution and the team was able to watch the drug bind

    to the tumor. This revealed the presence of the mutation

    and the effectiveness of the drug in blocking EGFR activity.

    Their revelation could be a big step in the treatment

    of NSCLC, explained Dr. Contessa, because a physician

    Radiobiology and Radiotherapy RESEARCH PROGRAM

    can see whether the drug is hitting the target at every site

    in the body. If tumors are not imaged by [11C]-erlotinib

    PET, doctors can consider using another tyrosine kinase

    inhibitor or irradiating the sites missed by the drug. The

    technique can also reveal when patients are developing

    resistance to a drug, either generally or at certain sites, and

    hence could benefit from changing or supplementing their

    therapies. “We hope this technique will help physicians

    make these decisions earlier, by giving them more

    information more quickly,” said Dr. Contessa.

    The next step, beginning now, is a Phase I trial with

    about two dozen patients at Smilow Cancer Hospital.

    That’s where the fourth member of the team comes in,

    medical oncologist Sarah Goldberg, MD, MPH, Assistant

    Professor of Medicine. “Dr. Goldberg is very interested in

    using this new technique to improve therapeutic decision-

    making for patients with EGFR mutations,” said Dr. Contessa.

    The team believes this technique can be used for other

    types of cancer. “There are other tumor sites with specific

    mutations, and we might be able to develop radiotracers

    that specifically interact with those mutant proteins to

    give us a way to image different types of tumors,” said Dr.

    Contessa. “We think we can start to personalize imaging

    to find mutations, and use imaging as a way to gauge

    responses to therapy.”

    16 Yale Cancer Center | Year in Review 2015

    Evan Morris, PhD and Joseph Contessa, MD, PhD

    Imaging the Epidermal Growth Factor Receptor

  • Texting to Check in on Patients

    Sarah Mougalian, MD

    Sarah Mougalian, MD , Assistant Professor

    of Medicine, specializes in treating patients with

    breast cancer. She found herself frustrated by one

    aspect of their care. Most breast cancers – about 75

    percent – are fueled by the hormone’s estrogen and/

    or progesterone. After patients with this type of breast

    cancer finish their primary treatments, typically surgery

    and perhaps followed by radiation and/or chemotherapy,

    they are prescribed a medication to regulate their

    hormones, with the goal of preventing a recurrence of

    cancer. The regimen sounds simple – one pill every day

    for five to ten years.

    Yet many of these patients do not follow this regimen.

    At Smilow Cancer Hospital, said Dr. Mougalian, it’s

    about 25 percent, and some studies have found non-

    adherence rates up to 50 percent. The reasons range from

    forgetfulness to unfilled prescriptions to intolerance of the

    medication’s side effects. Failure to take the medication or

    to complete the long-term therapy may put these patients

    at higher risk of recurrence. Hence Dr. Mougalian’s

    frustration, which has led to an innovative solution.

    Many of Dr. Mougalian’s patients are young women,

    and she noticed that when she entered an examining

    room, they often were texting on their smart phones. “So

    we thought about creating a text messaging system to help

    people remember to take their medication and to identify

    any problems early,” explained Dr. Mougalian. “It can be

    three to six months between appointments, a long time

    to wait if you have questions. In some cases, people stop

    taking their medication and we don’t even know it.”

    Dr. Mougalian collaborated with the technology

    company CircleLink Health to design a new idea in

    cancer treatment – a pilot study built around automated

    text messages. She enrolled 100 patients for three months.

    All of them received a daily text asking if they had taken

    their medication. They type “Y” for yes or “N” for no.

    Anyone who didn’t reply or who texted N for three

    straight days or six times within 30 days received a phone

    call from a triage nurse to find out what was going on.

    “This is an effort to identify people who are at risk of

    discontinuing their medication, and to try to address it in

    real time,” said Dr. Mougalian.

    Every patient also got a weekly text asking whether they

    were experiencing any of the listed side effects, and if so, to

    rate them on a scale of severity from one to nine. Anyone

    who reported severe side effects received a phone call to

    discuss behavioral modifications or were offered a sooner

    appointment with their physician. Patients could also free-

    text non-listed side effects.

    Finally, patients got one text per month asking if

    Cancer Prevention and Control RESEARCH PROGRAM

    anything was preventing them from continuing their

    medication, such as side effects or finances.

    Dr. Mougalian wanted to know whether patients

    would be irritated by the daily texts, or if they would

    develop “alert fatigue” and delete the reminder without

    replying. Only one patient complained. “Lots of patients

    say they like it, because they feel like somebody is

    looking out for them,” said Dr. Mougalian, “and it’s also

    an accountability tool, because they know someone is

    checking up on them.”

    The text system also seemed to improve adherence:

    only five of the 100 patients stopped taking their

    medication (four due to side effects), a much higher rate

    of adherence than is typical. Dr. Mougalian cautions

    that this finding is preliminary. She hopes to test it in a

    long-term randomized study and has applied for funding

    to do a five-year investigation with 400 patients, half of

    them using the text system and the other half not, as a

    control group.

    “The ultimate goal,” she said, “is to see whether better

    adherence correlates to better survival in the long run.”

    19yalecancercenter.org | Yale Cancer Center

    “The ultimate goal is to see whether

    better adherence correlates to better

    survival in the long run.”

  • Not every lab discovery leads directly to

    a clinical application, but progress in cancer

    care depends on insights from basic science into the

    fundamental mechanisms of biology.

    A recent paper in the journal Cell offers a good

    example. Scientists from Yale Cancer Center and

    elsewhere have been studying links between lymphoma

    and a biological mechanism called V(D)J recombination.

    This mechanism recombines broken pieces of DNA,

    and is essential for the development of T cells and B

    cells, lymphocytes that form the adaptive immune

    system. Yet this process of genetic recombination is

    also risky. The paper’s lead author, Grace Teng, PhD, a

    postdoctoral fellow and Associate Research Scientist in

    Immunobiology, wrote that these developing T and B cells

    “perch on the edge of genomic instability.”

    “When broken DNA ends are flopping around in the

    cell, they can become joined in a haphazard fashion,”

    explained Dr. Teng. “A lot of cancers are associated

    with these erroneous repair processes. The fact that

    lymphocytes have to go through programmed DNA

    damage during development makes them particularly

    susceptible to errors. That’s why lots of lymphomas stem

    from B and T cells.”

    In short, the mechanism that creates our adaptive

    immune system also exposes us to the risk of lymphoma.

    “New lymphocytes are being generated in our bodies at

    a tremendous rate every day,” said another of the paper’s

    authors, David Schatz, PhD, Professor of Immunobiology

    and of Molecular Biophysics and Biochemistry, “and

    the V(D)J recombination process happens hundreds

    of millions, if not billions of times per day. So the risk is

    ongoing and chronic, and even an extremely low error rate

    gives you a significant risk.”

    Dr. Teng’s research also uncovered an unexpected

    wrinkle in V(D)J recombination: breaks of DNA in the

    “wrong” places. In V(D)J recombination, DNA is cut by

    two proteins called recombination activating genes 1 and 2

    (RAG1 and RAG2), which Dr. Schatz discovered 25 years

    ago. They were thought to cut DNA in a small, limited part

    of the genome, as a way of protecting the rest of the genome

    from flawed cuts. But Dr. Teng and her colleagues found

    RAG1 and RAG2 in thousands of off-target sites. “That

    seemed to constitute a much broader, more significant

    threat than previously suspected,” said Dr. Schatz.

    Dr. Teng and colleagues knew that the RAG

    complex prefers to sever DNA at specific spots called

    Recombination Signal Sequences (RSSs), which are

    abundant in the normal cutting sites. Researchers found

    that in the off-target sites, RSSs are significantly depleted.

    Cancer Immunology RESEARCH PROGRAM

    That means fewer places for RAG to cut, which lessens the

    risk of incorrectly refitted strands of DNA.

    “You end up with two different compartments of the

    genome,” said Dr. Teng, “one enriched with RSSs, and

    another where there’s not much of the DNA sequence for

    RAG to cut.” This compartmentalization seems to protect

    the genome from inappropriate cuts in the off-target sites.

    That begs another question: why didn’t evolution

    remove this bug from the system? Dr. Schatz points out

    that RAG1 and RAG2, with the threat of lymphoma they

    carry, have been present during vertebrate evolution for

    hundreds of millions of years. The depletion of RSSs at off-

    target sites might be evolution’s way of putting the threat

    on the back-burner.

    “Cancer is generally a disease of old age,” said Dr.

    Schatz, “and hence lymphomas that would kill people at

    50 or 60 would have been a very weak evolutionary force

    for most of human evolution. I suspect that’s why this

    hasn’t been completely selected against.”

    These discoveries might someday help physicians

    predict which parts of the genome are at greatest risk

    of lymphoma. “As basic scientists,” said Dr. Schatz, “we’re

    not looking at how the immune system is interacting

    with tumors, but at what gave rise to the tumors in the

    first place.”

    20 Yale Cancer Center | Year in Review 2015

    Grace Teng, PhD and David Schatz, PhD

    The Immune System and the Risk of Lymphoma

  • Jorge Galan, PhD, DVM

    The Links Between Typhoid, Bacteria, and Cancer

    A discovery by a Yale Cancer Center

    researcher promises to beget a vaccine against one of

    the world’s most deadly diseases, typhoid fever. This

    breakthrough is also likely to cause two important side

    effects: a decline in the incidence of gallbladder cancer,

    and more research on the links between bacterial

    pathogens and cancer.

    Typhoid fever is one of the scourges of public health,

    sickening more than 20 million people every year and

    killing 200,000, almost all of them in undeveloped

    countries, where it is spread by contaminated food

    or water, usually via the feces of infected people. The

    infectious cause of typhoid has been known for more than

    a century, a bacterium named Salmonella Typhi. But the

    bacteria’s mechanism to cause disease has been a mystery,

    preventing the development of a knock-out vaccine, one

    of the holy grails of public health.

    That may soon change. A team led by Jorge Galan,

    PhD, DVM, Lucille P. Markey Professor of Microbial

    Pathogenesis and Cell Biology, and Chair of Microbial

    Pathogenesis, has identified the basis for Salmonella

    Typhi’s unique virulence properties. Dr. Galan’s

    team identified “typhoid toxin,” which is produced

    by Salmonella Typhi and in experimental animals, can

    reproduce most of the symptoms of typhoid fever.

    Dr. Galan’s team has also solved the atomic structure

    of “typhoid toxin,” which should make possible the

    development of small molecule inhibitors to block the

    infection, as well as a vaccine that could potentially

    eradicate typhoid fever.

    “This discovery turned the disease upside down,” said

    Dr. Galan. “Now we know why S. Typhi causes typhoid

    fever, and we can create a vaccine against it. That’s the main

    impact.” The Bill & Melinda Gates Foundation has put Dr.

    Galan’s breakthrough onto a fast track towards a vaccine.

    The discovery also provides a molecular explanation for

    the epidemiological link between typhoid and gallbladder

    cancer. Many survivors of typhoid become asymptomatic

    carriers of S. Typhi, and hold the bacteria in the gallbladder.

    “We discovered that S. Typhi encodes what is essentially

    a genotoxin – a toxin that damages the genome’s DNA,”

    said Dr. Galan. “So the tumorigenesis is likely to be related

    to the toxin’s ability to damage the DNA. When the DNA

    is mis-repaired, mutations are created, providing the

    basis for the development of cancer. So a vaccine against

    typhoid fever should also prevent gallbladder cancer.”

    That would be significant in the developed world as

    well. About 10,000 new cases of gallbladder cancer are

    diagnosed in the United States each year, and 4,000 people

    die from it.

    Virus and Other Infection-associated Cancers RESEARCH PROGRAM

    Dr. Galan believes that the connections between cancer

    and infections, especially bacterial infections, deserve more

    attention. “This story of ours is one more example. Cancer

    researchers sometimes forget that 20 percent of known

    cancers are caused by an infectious organism,” he said,

    “and that’s probably a gross underestimate, because it only

    captures instances where the microorganism is the direct

    cause – for example, in the case of human papillomavirus

    (HPV) and cervical cancer, or the bacterium Helicobacter

    pylori and stomach cancer.”

    In far more instances, he added, bacteria and viruses

    are most likely predisposing factors that essentially tilt the

    scale leading to cancer. “There are many cases of bacteria

    that produce genotoxic agents, particularly bacteria that

    hang around in the gut, and the thought is that some of

    these bacteria could be predisposing factors, for example,

    for colon cancer. It is also widely believed that the resident

    microbiota in our body most likely plays an important role

    in predisposing to certain types of cancer. This is a frontier

    that we’re just beginning to explore.”

    It is also the impetus behind the name change of the

    Research Program from Molecular Virology to Virus

    and Other Infection-associated Cancers. “The idea is to

    broaden the focus, to think about other microorganisms as

    cancer causing agents and not just viruses,” said Dr. Galan.

    23yalecancercenter.org | Yale Cancer Center

  • Cells constantly produce new proteins, which

    wear themselves out doing cellular labor. Spent proteins

    are tagged for removal by a maintenance protein called

    ubiquitin, and then sent to the proteasome, the body’s

    equivalent of the glue factory. There, the old proteins

    are broken down and discarded. This process is called

    protein degradation.

    Craig M. Crews, PhD, Lewis B. Cullman Professor of

    Molecular, Cellular and Developmental Biology, has been

    studying this process for years, looking for ways to control

    it with drug-like molecules that target cancer-causing

    proteins. His lab’s first foray into this field led to the drug

    Kyprolis® (carfilzomib), approved in 2012 for use against

    multiple myeloma. Kyprolis® is a proteasome inhibitor

    that prevents the degradation of protein in cancer cells,

    causing a toxic build-up that kills them.

    Now Dr. Crews and his colleagues have developed

    a different line of attack. “Instead of blocking protein

    degradation,” said Dr. Crews, “we’re inducing it. We’ve

    developed a drug strategy whereby the drug enters the cell,

    seeks out and binds to rogue cancer-causing proteins, and

    drags them to the proteasome for degradation. It’s a seek

    and destroy approach.” These drug-like molecules, called

    PROTACs (Proteolysis-Targeting Chimeras), were first

    reported by Dr. Crews in 2001.

    Most targeted cancer drugs are inhibitors that bind

    to receptors and block the mutant proteins that cause

    cancer. “But when the drug leaves the system, the protein

    can start working again,” Dr. Crews explained. “So, to get

    the clinical benefit, you need to maintain a high level of

    the drug in the body. What we’re doing is different. It’s

    not binding and gumming up the process, it’s tricking

    the cell’s own quality control machinery to destroy rogue

    proteins. It’s also permanent. And the drug survives to

    eat its way through a protein population, so in theory,

    one needs less of our drug. This approach should greatly

    reduce the side effects that accompany high dosing in the

    inhibitor paradigm.”

    Another strength is the strategy’s broad reach. Dr.

    Crews points out that each cell contains an estimated

    20,000 different proteins, but only about 25 percent

    are potentially vulnerable to inhibitors, which work by

    binding receptors or blocking enzymatic functions. That

    leaves the great majority of proteins untouched, such as

    scaffolding proteins and transcription factors. Since Dr.

    Crews’ approach doesn’t depend on inhibition, it should

    work against this so-called ‘undruggable’ majority.

    The science of tagging and eliminating oncogenic

    proteins is new, and so is the technology that makes

    it possible: a new class of small-molecule PROTACs

    Developmental Therapeutics RESEARCH PROGRAM

    designed by Dr. Crews and his lab, a collection of

    cell biologists, biochemists, medical chemists, and

    pharmacologists. Dr. Crews believes that small-molecule

    PROTACs open up wide new possibilities for cancer

    therapies. He has started a company called Arvinas to

    explore them and to develop the technology further.

    For now, Dr. Crews and his partners are interested in

    developing PROTACs for leukemias, lymphomas, prostate

    cancer, breast cancer, and lung cancer. “But this approach

    doesn’t really have any limitations with respect to cancer

    type,” said Dr. Crews. “It’s a platform technology, so the

    learning around developing one drug could be applied to

    address the oncogenic proteins that cause different cancers

    in different tissues.” He hopes that clinical trials for

    prostate cancer will begin sometime next year.

    The National Institutes of Health (NIH) noticed this

    breakthrough and Dr. Crews was recently selected for the

    inaugural class of a new program at the NIH. He received

    an Outstanding Investigator Award and funding of $1

    million for each of the next seven years.

    “I’m very fortunate that for the last 20 years my lab has

    worked right at the interface of chemistry and biology.

    It allows me to recognize the important questions in

    biology and allows my lab to then go out and tackle those

    problems using chemical approaches,” said Dr. Crews.

    24 Yale Cancer Center | Year in Review 2015

    Molecules That Seek and Destroy Mutant Proteins

    Craig M. Crews, PhD

  • Michael Krauthammer, MD, PhD

    Bioinformatics Boosts Cancer Research

    27yalecancercenter.org | Yale Cancer Center

    Biomedical informatics is changing our

    understanding and treatment of cancer. That was recently

    illustrated when a team of Yale Cancer Center scientists

    used molecular sequencing and computational biology to

    reveal startling new information about melanoma.

    The team analyzed exome data from 213 melanoma

    patients, the second largest such screening ever done.

    “We were looking for a better understanding of all the

    types of mutations that must happen for melanoma to

    occur,” said Michael Krauthammer, MD, PhD, Associate

    Professor of Pathology.

    They found several surprises. Scientists already knew

    that most melanomas stem from mutations in either the

    BRAF gene (about 50 percent) or the NRAS gene (20 to

    30 percent), but the mutations that caused the remaining

    20 to 30 percent were unclear. Researchers did know that

    some melanomas contained mutations in the NF1 gene,

    but NF1’s role wasn’t well defined.

    The Yale screen, conducted as part of the Yale SPORE

    in Skin Cancer, showed that mutations of NF1 account

    for 10 to 15 percent of melanomas, making them the

    third most important driver of the cancer. Further, NF1

    mutations help activate the MAPK pathway, the same one

    activated by oncogenic mutations in BRAF and NRAS.

    “This moves patients with an NF1 mutant melanoma

    into a much better understood category,” explained

    Dr. Krauthammer, “particularly because it’s acting on

    a pathway for which we already have multiple drugs

    available. We didn’t have this knowledge before.”

    There were other surprises. First, the genome of NF1

    patients was much more mutated than those of other

    melanoma patients. Second, patients in the NF1 subgroup

    were also much older.

    “That puzzled us,” said Dr. Krauthammer. “Why

    would they have so many more mutations – thousands

    of them – and be older?” The researchers noticed that

    NF1 mutations didn’t seem to be strong activators of

    MAPK. “That led to the interesting hypothesis that maybe

    these NF1 melanomas have to reside in the body longer

    and acquire more mutations to become as active and

    malignant as a BRAF or NRAF melanoma.”

    The research team discovered that NF1 melanomas were

    acquiring additional mutations from a parallel disease,

    RASopathies. RASopathies are developmental disorders,

    such as Noonan syndrome and Neurofibromatosis, caused

    by germline (inherited) genetic mutations, as distinct

    from somatic mutations in cancer, which happen after

    conception and can’t be inherited. RASopathies were not

    known to be related to melanoma, but they do activate

    the MAPK pathway – the same one used by NF1. Dr.

    Cancer Genomics, Genetics, and Epigenetics RESEARCH PROGRAM

    Krauthammer and his team found that the mutations

    in RASopathies were identical to those in mutated

    NF1. Further analysis suggested that NF1 mutations by

    themselves are too weak to cause melanoma, but when

    joined with RASopathy mutations, the combination may

    activate the MAPK pathway for melanoma.

    These findings began with the analysis of 20,000 genes

    for the mutations most important to melanoma. They

    have narrowed the list to about 100 genes – still a sizable

    number. “Melanoma has so many more mutations than

    other cancers,” said Dr. Krauthammer, “but we were

    able to identify a key subset of genes that we are going

    to sequence across a large cohort of melanoma patients

    that are treated at Yale. That will give us a big hint

    about what his or her response will be to the targeted

    therapies already in use against melanoma, or possibly to

    the new immunotherapies.”

    The fact that so many mutated genes are implicated

    in melanoma is altering our understanding of cancer.

    “Suddenly it’s less about whether a disease is a

    developmental delay, or a skin cancer, or a blood cancer,”

    he added, “and more about the underlying molecular

    mechanism. That’s why bioinformatics is so useful in

    modern cancer research – to analyze a lot of data, recognize

    patterns, and establish links to mutations in other diseases.”

  • “There is a bit of a misplaced emphasis in

    the clinic and in clinical science as to how we think

    about targeting cancer,” said Andre Levchenko, PhD,

    John C. Malone Professor of Biomedical Engineering and

    Director of the Yale Systems Biology Institute. “There

    has been a strong focus on controlling primary tumor

    growth, even though we know that more than 90 percent

    of cancer deaths occur because of invasive cell behavior,

    or metastasis. The main problem isn’t the growth of

    the primary tumor, it’s the ability of cancer cells to

    change their behavior to invasive migration and travel to

    secondary sites.”

    The emphasis on tumor growth, he added, stems partly

    from the difficulty of studying the causes and engines of

    cancer cell migration. That’s changing, thanks to new tools

    and technologies, some of them unique to Dr. Levchenko’s

    lab, where custom-built devices and new methods of

    analysis are allowing researchers to trace the movement

    of cancer cells, the cues that incite their invasion, and the

    signals that guide them to new locations during metastasis.

    “Their circuits get rewired,” said Dr. Levchenko,

    “so instead of staying put, they start moving toward

    this big highway system, the bloodstream.” Eventually

    they navigate toward tissues very different from their

    native environments, constantly adjusting to different

    circumstances, and colonize. “The process is very complex

    and not well understood,” said Dr. Levchenko, “but it’s a

    systems problem, so we need to use systems approaches.”

    His lab and other groups at the Systems Biology

    Institute, which he has been leading for the past two

    years on Yale’s West Campus, are trying to understand

    what transforms site-specific tumor cells into a far-

    ranging invasive army. What networks of molecules

    mediate this switch? How do invasive cells communicate

    and coordinate? Which neighboring cells resist? Which

    collaborate with the invaders? How do cancer cells

    develop new behaviors and change the behaviors of

    neighboring cells?

    Many of the answers lie within the complex system

    of signaling networks, the fundamental interest of Dr.

    Levchenko’s lab and the Institute. In cancer, normal

    communication between cells gets misinterpreted,

    disrupted, or corrupted. “If we can understand the

    signaling mechanisms,” he said, “we can develop new

    therapies that prevent invasive spread – for instance, by

    rewiring these networks. There’s already a lot of activity to

    generate novel therapeutics to do that, and we are working

    with others to determine the fastest translational path for

    clinical applications. That’s the ultimate goal.”

    They are concentrating on what Dr. Levchenko calls

    Signal Transduction RESEARCH PROGRAM

    “the champions of invasive behavior” – glioblastoma

    and melanoma, two of the most deadly and fast-moving

    cancers, and also breast cancer, another aggressive

    malignancy. A recent paper described how individual

    metastasizing breast cancer cells become generally

    exploratory, searching for the bloodstream, and then,

    in response to a cue, suddenly band together in one

    direction, as if they have received their orders of invasion.

    “As soon as there is a clear indication of where they want

    to go,” explained Dr. Levchenko, “they begin moving

    almost in goose step with each other.”

    He and his colleagues were able to collect this

    extraordinary information by building devices that can

    track the path of a single cell and its changing behaviors in

    response to cues and signaling networks. These tools give

    the scientists novel ways to investigate and analyze a single

    cell, looking at every gene, at the metabolizing proteins,

    and other factors.

    He believes that understanding the complexity of

    signaling networks and the process of metastasis demands

    a multidisciplinary approach. The teams he has put

    together at his lab and institute include researchers of

    diverse disciplines: evolutionary biologists, synthetic

    biologists, and systems biologists, but also physicists,

    mathematical modelers, and biomedical engineers.

    28 Yale Cancer Center | Year in Review 2015

    Decoding the Signals of Metastasis

    Andre Levchenko, PhD

  • Yale Cancer Center

    Peter G. Schulam, MD, PhDInterim Director

    Daniel C. DiMaio, MD, PhDDeputy Director

    Kevin Vest, MBA, FACHEDeputy Director, Finance and Administration

    Mark A. Lemmon, PhDCo-Director, Cancer Biology Institute

    Robert Garofalo, PhDAssociate Director, Research Affairs

    Roy S. Herbst, MD, PhDAssociate Director, Translational Science

    Howard S. Hochster, MDAssociate Director, Clinical Sciences

    Melinda L. Irwin, PhD, MPHAssociate Director, Population Sciences

    Patricia M. LoRusso, DOAssociate Director, Innovative Medicine

    David F. Stern, PhDAssociate Director, Shared Resources

    Joann B. Sweasy, PhDAssociate Director, Basic Science

    Anees B. Chagpar, MDAssistant Director, Global Oncology

    Beth Jones, PhD, MPHAssistant Director, Diversity and Health Equity

    Harriet Kluger, MDAssistant Director, Education and Training

    Yale Cancer Center Research Programs

    Cancer Genomics, Genetics, and EpigeneticsMarcus W. Bosenberg, MD, PhDLajos Pusztai, MD, DPhil

    Cancer ImmunologyLieping Chen, MD, PhDMadhav V. Dhodapkar, MD, PhD, MBBS

    Cancer Prevention and ControlMelinda L. Irwin, PhDXiaomei Ma, PhD

    Developmental TherapeuticsKaren S. Anderson, PhDBarbara A. Burtness, MD

    Virus and Other Infection-associated CancersWalther H. Mothes, PhDWendell G. Yarbrough, MD

    Yale Cancer Center Shared Resources

    Biostatistics and BioinformaticsPeter Peduzzi, PhD

    Cesium IrradiatorRavinder Nath, PhD

    Clinical Research ServicesHoward S. Hochster, MD

    Flow CytometryAnn Haberman, PhD

    Pathology Tissue ServicesDavid Rimm, MD, PhD

    Rapid Case AscertainmentRajni Mehta, MPH

    Yale Center for Genome AnalysisShrikant Mane, PhD

    Yale Center for Molecular DiscoveryCraig Crews, PhD

    30 Yale Cancer Center | Year in Review 2015

    Yale Cancer Center Leadership

    Radiobiology and RadiotherapyPeter M. Glazer, MD, PhDJoann B. Sweasy, PhD

    Signal TransductionDaniel P. Petrylak, MDDavid F. Stern, PhD

    Gary Kupfer, MDAssistant Director, Pediatrics

    Ruth McCorkle, RN, PhDAssistant Director, Psychosocial Oncology

    Peter G. Schulam, MD, PhDAssistant Director, Surgery

    Andrea Silber, MDAssistant Clinical Director, Diversity and Health Equity

    Jeffrey Sklar, MD, PhDAssistant Director, Pathology & Tissue Acquisition Services

    Edward Snyder, MDAssistant Director, Membership

    31yalecancercenter.org | Yale Cancer Center

    Smilow Cancer Hospital at Yale-New Haven

    Peter G. Schulam, MD, PhDPhysician-in-Chief (Interim)

    Abe LopmanSenior Vice President, Operations and Executive Director

    Rogerio C. Lilenbaum, MDChief Medical Officer

    Catherine Lyons, RN, MSExecutive Director, Smilow Patient Care Services

    Kerin B. Adelson, MDChief Quality OfficerDeputy Chief Medical Officer

    Anne Chiang, MD, PhDChief Network OfficerDeputy Chief Medical Officer

    Benjamin L. Judson, MDMedical Director, Ambulatory ClinicsDeputy Chief Medical Officer

    Michael D. Loftus Vice President, Financial Operations

    Arthur LemayExecutive Director, Smilow Cancer Network

    Yale Cancer Center and Smilow Cancer Hospital at Yale-New Haven have joined forces to create Closer to Free, a fund that provides essential financial support for break-through cancer research and compassionate patient care. Learn More >> www.giveclosertofree.org

    closer to freegive

    Be part of our mission to bring the world Closer to Free.

    Smilow Cancer Hospital Clinical Programs

    Brain Tumor Clinical Program Leader:Joachim M. Baehring, MDDisease Aligned Research Team Leader:Kevin P. Becker, MD, PhD

    Breast Cancer Clinical Program Leader:Anees B. Chagpar, MDDisease Aligned Research Team Leader:Lajos Pusztai, MD, DPhil

    Endocrine CancersClinical Program and Disease Aligned Research Team Leader:Tobias Carling, MD, PhD

    Gastrointestinal CancersClinical Program and Disease Aligned Research Team Leader:Howard S. Hochster, MD

    Gynecologic Cancers Clinical Program Leader:Peter Schwartz, MDDisease Aligned Research Team Leader:Alessandro D. Santin, MD

    Head and Neck Cancers Clinical Program Leader: Wendell G. Yarbrough, MDDisease Aligned Research Team Leader:Barbara A. Burtness, MD

    Hematology Clinical Program Leader: Steven D. Gore, MDDisease Aligned Research Team Leader:Madhav V. Dhodapkar, MD, PhD, MBBS

    Melanoma Clinical Program Leader:Stephan Ariyan, MDDisease Aligned Research Team Leader:Mario Sznol, MD

    Phase IClinical Program Leader:Patricia M. LoRusso, DO Disease Aligned Research Team Leader:Joseph Paul Eder, MD

    Prostate and Urologic Cancers Clinical Program Leader: Peter G. Schulam, MD, PhDDisease Aligned Research Team Leader:Daniel P. Petrylak, MD

    SarcomaClinical Program Leader:Gary E. Friedlaender, MDDisease Aligned Research Team Leader:Dieter M. Lindskog, MD

    Therapeutic Radiology Clinical Program Leader:Lynn D. Wilson, MD, MPHDisease Aligned Research Team Leader:Roy H. Decker, MD, PhD

    Thoracic Oncology Clinical Program Leader:Frank C. Detterbeck, MDDisease Aligned Research Team Leader:Roy S. Herbst, MD, PhD

    Pediatric Hematology and OncologyClinical Program and Disease Aligned Research Team Leader:Gary Kupfer, MD

  • Allen Everett BaleLinda M. BartoshukSusan J. BasergaLauren Paige BlairJean L. BologniaMarcus W. BosenbergDemetrios BraddockTobias CarlingNancy CarrascoSidi ChenKeith Adam ChoateLynn CooleyJose CostaBernard G. ForgetAlan GarenMark B. GersteinAntonio J. GiraldezMurat GunelShangqin GuoRuth HalabanStephanie HaleneShilpa HattangadiChristos HatzisErin Wysong HofstatterJosephine J. HohNatalia B. IvanovaSamuel G. KatzSajid A. KhanKenneth Kay KiddYuval KlugerWilliam H. KonigsbergDiane S. Krause

    Rossitza LazovaDavid J. LeffellPeter LengyelPeining LiRichard P. LiftonHaifan LinXavier LlorJun LuShrikant M. ManeMiguel A. MaterinJames Michael McGrathKarla NeugebauerJames P. NoonanManoj PillaiManju PrasadLajos PusztaiPeter E. SchwartzEmre U. SeliGerald S. ShadelJeffrey L. SklarMatthew Perry StroutHugh S. TaylorJeffrey TownsendRobert UdelsmanScott Donald WeatherbeeSherman Morton WeissmanAndrew Zhuo XiaoMina LuQing XuTian XuQin YanHongyu Zhao

    Stephan AriyanPhilip William AskenaseKevin Patrick BeckerJeffrey R. BenderAlfred L. M. BothwellRichard BucalaLieping ChenOscar Rene ColegioJoseph Edgar CraftPeter CresswellKavita DhodapkarMadhav V. DhodapkarRichard L. EdelsonBrinda EmuRichard A. FlavellFrancine M. FossMichael GirardiEarl John GlusacAnn M. HabermanDavid HaflerDouglas John HanlonPaula B. KavathasSteven H. KleinsteinSmita KrishnaswamyMark Joseph MamulaJennifer Madison McNiffRuslan M. MedzhitovEric R. F. MeffreDeepak NarayanJoao P. PereiraJordan Stuart PoberCarla Vanina RothlinNancy Hartman RuddleKurt SchalperDavid G. SchatzStuart Evan SeropianBrian Richard SmithEdward Leonard SnyderMario SznolRobert E. TigelaarJun Wang

    Kerin Bess AdelsonSteven L. BernsteinElizabeth H. BradleyBrenda CartmelAnees B. ChagparElizabeth Brooks ClausAmy Joan DavidoffNicole Cardello DezielRobert DubrowLeah McArthur FerrucciLisa FucitoBonnie Elyssa Gould RothbergCary P. GrossCaitlin Elizabeth HansenTheodore R. HolfordScott Frederick Huntington Melinda Liggett IrwinBeth A. JonesManisha Juthani-Mehta Nina S. Kadan-LottickJennifer M. KapoBrigid KilleleaAnthony W. KimTish KnobfSuchitra Krishnan-SarinStephanie Lynn KweiDonald R. LanninJames Mark Lazenby

    Steven B. LederHaiqun LinLingeng LuShuangge Steven MaXiaomei MaAsher Michael MarksRuth McCorkleSherry McKeeRajni Lynn MehtaSarah Schellhorn MougalianLinda M. NiccolaiMarcella Nunez-SmithStephanie Samples O’MalleyJonathan Thomas PuchalskiElena RatnerHarvey A. RischPeter SaloveyTara SanftDena J. Schulman-GreenDave SellsFatma M. SheblSangini S. ShethAndrea Lynn Maria SilberMehmet SofuogluShiyi WangYawei ZhangYong Zhu

    32 Yale Cancer Center | Year in Review 2015

    Yale Cancer Center Membership

    Cancer Epigenetics, Genetics, and Genomics

    Cancer Immunology Cancer Prevention and Control

    33yalecancercenter.org | Yale Cancer Center

    Janet L. BrandsmaDaniel C. DiMaioAyman Sayed El-GuindyJorge E. GalanAndrew GoodmanStanley David HudnallNatalia IssaevaAkiko IwasakiBenjamin L. JudsonSusan M. KaechMichael J KozalMartin Alexander KriegelPriti KumarBrett D. LindenbachRobert E. MeansI. George MillerKathryn Miller-JensenWalther H. MothesElijah Paintsil Noah Wolcott PalmAnna Marie PyleMichael RobekJohn K. RoseAlessandro D. SantinChristian SchliekerJoan A. SteitzRichard E. SuttonPeter John TattersallAnthony N. Van den PolYong XiongWendell Gray Yarbrough

    Ranjit S. BindraDaniel J. BoffaDouglas E. BrashDavid Joel CarlsonRichard E. CarsonSandy ChangZhe (Jay) ChenVeronica Lok Sea ChiangJohn W. ColbergJoseph N. ContessaShari DamastRoy H. DeckerJun DengFrancesco DErricoFrank C. DetterbeckJames S. DuncanSuzanne B. EvansPeter Michael GlazerFanqing GuoJames E. HansenHoby Patrick HetheringtonSusan A HigginsZain A. HusainFahmeed HyderRyan B. JensenMegan C. KingGary KupferWu LiuK. Brooks LowMeena Savur MoranEvan Daniel MorrisRavinder NathAbhijit A. PatelRichard E. PeschelKenneth B. RobertsFaye A. RogersPeter SchulamYung H. SonPatrick SungJoann Balazs SweasyLynn D. WilsonSandra L. WolinJames Byunghoon YuZhong Yun

    Anton M. BennettKenneth F. BlountTitus BoggonDavid A. CalderwoodLloyd Garnet CantleyToby C. ChaiPietro De CamilliGary Vincent DesirMichael P. DiGiovannaRong FanKathryn M. FergusonJohn P. GeibelSourav GhoshValentina GrecoJaime GrutzendlerMark W. HochstrasserValerie HorsleyMichael E. HurwitzKarl L. InsognaRichard Glenn KibbeyJoseph W. KimAnthony J. KoleskeMichael Oliver KrauthammerTuKiet T. LamMark A. LemmonAndre LevchenkoJoseph Anthony MadriWang MinJon Stanley MorrowPeggy Myung Michael H. NathansonDon X. NguyenDaniel PetrylakKaterina PolitiDavid L. RimmJoseph SchlessingerMartin A. SchwartzDavid F. SternYajaira SuarezDerek K. ToomreBenjamin E. TurkNarendra WajapeyeeRobert Martin WeissKenneth R. WilliamsDan WuJohn Joseph WysolmerskiXiaoyong Yang

    Maysa Mahmoud Abu-KhalafKaren S. AndersonMasoud AzodiJoachim M. BaehringAarti Khushal BhatiaDebra Schwab BrandtRonald R. BreakerBarbara Ann BurtnessCharles H. ChaHerta H. ChaoYung-Chi ChengAnne ChiangJennifer Nam ChoiGina G. ChungJason Michael CrawfordCraig M. CrewsHenk De FeyterHari Anant DeshpandeVincent T. DeVitaJoseph Paul EderBarbara E. EhrlichJonathan A. EllmanDonald Max EngelmanTarek FahmyLeonard Raymond FarberJames J. FarrellJean-Francois GeschwindScott Nicholas GettingerSarah B. GoldbergSteven D. GoreYa HaDale HanRoy S. HerbstSeth B. HerzonHoward S. HochsterMichael Edwin HodsdonNina Ruth HorowitzWilliam L. Jorgensen

    Developmental Therapeutics Virus and Other Infection-associated Cancers

    Radiology and Radiotherapy Signal Transduction

    Juliane M. JuergensmeierPatrick A. KenneyHarriet M. KlugerJaseok KooJill LacyJia LiRogerio C. LilenbaumDieter M. LindskogElias LolisPatricia LoRussoScott J. MillerJennifer MoliternoGil G. MorNatalia NeparidzeTerri Lynn ParkerPasquale PatrizioPeter Natale PeduzziAndrew J. PhillipsJoseph Massa PiepmeierNikolai Alexandrovich PodoltsevThomas PrebetLynne J. ReganJohn David RobertsMichal Gillian RoseThomas J. RutherfordW. Mark SaltzmanAlan Clayton SartorelliClarence Takashi SasakiAlanna SchepartzWilliam C. SessaBrian Matthew ShuchDavid Adam SpiegelPreston SprenkleStacey M. SteinSeyedtaghi (Shervin) TakyarVasilis VasiliouDaniel ZeltermanJiangbing Zhou